EP3411052B1 - Utilisation de communautés microbiennes pour la santé humaine et animale - Google Patents

Utilisation de communautés microbiennes pour la santé humaine et animale Download PDF

Info

Publication number
EP3411052B1
EP3411052B1 EP17703726.4A EP17703726A EP3411052B1 EP 3411052 B1 EP3411052 B1 EP 3411052B1 EP 17703726 A EP17703726 A EP 17703726A EP 3411052 B1 EP3411052 B1 EP 3411052B1
Authority
EP
European Patent Office
Prior art keywords
composition
bacteria
gut
species
intestinal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP17703726.4A
Other languages
German (de)
English (en)
Other versions
EP3411052B8 (fr
EP3411052A1 (fr
Inventor
Sam Possemiers
Massimo Marzorati
Tom Van De Wiele
Ilse SCHEIRLINCK
Pieter VAN DEN ABEELE
Selin BOLCA
Davide GOTTARDI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universiteit Gent
Microbial Resource Management Health NV MRM Health
Original Assignee
Universiteit Gent
Prodigest BVBA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to DK19212983.1T priority Critical patent/DK3639834T3/da
Application filed by Universiteit Gent, Prodigest BVBA filed Critical Universiteit Gent
Priority to EP23182773.4A priority patent/EP4257194A3/fr
Priority to EP19212983.1A priority patent/EP3639834B1/fr
Priority to PL17703726T priority patent/PL3411052T3/pl
Priority to SI201730441T priority patent/SI3411052T1/sl
Priority to RS20201197A priority patent/RS60918B1/sr
Publication of EP3411052A1 publication Critical patent/EP3411052A1/fr
Application granted granted Critical
Publication of EP3411052B1 publication Critical patent/EP3411052B1/fr
Priority to HRP20201561TT priority patent/HRP20201561T1/hr
Publication of EP3411052B8 publication Critical patent/EP3411052B8/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/745Bifidobacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0031Rectum, anus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/26Means for regulation, monitoring, measurement or control, e.g. flow regulation of pH
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/30Means for regulation, monitoring, measurement or control, e.g. flow regulation of concentration
    • C12M41/34Means for regulation, monitoring, measurement or control, e.g. flow regulation of concentration of gas
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/11Lactobacillus
    • A23V2400/169Plantarum
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/51Bifidobacterium
    • A23V2400/513Adolescentes
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/51Bifidobacterium
    • A23V2400/533Longum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K2035/11Medicinal preparations comprising living procariotic cells
    • A61K2035/115Probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a mixture of bacteria belonging to at least 6 or 7 different and specific bacterial species preferably for use to prevent or treat gastro-intestinal disorders. More preferably, said mixture of bacteria are grown together in a fermenter prior to administering said mixture to a subject in order to prevent or treat said disorder.
  • the human and animal gut ecosystem consists of a variety of different habitats and metabolic niches that are colonized by the so-called microbiota that contain more than 10 11 micro-organisms per gram wet weight of contents, predominantly anaerobes (Macfarlane & Macfarlane, 1997). It is nowadays well-recognized that the human or animal gut microbiome plays a crucial role in human health and well-being by contributing to energy harvest, modulating the immune system and establishing colonization resistance against opportunistic pathogens (Fuller & Gibson, 1997; Cummings & Macfarlane, 1997). Evidence exists that the interaction of bacteria and their metabolites with the mucus layer and/or with the intestinal gut wall is important (Barnett et al. 2012).
  • Dysbiosis is characterized by moderate or severe disruptions in the normal gut microbiome composition, thereby causing the lack of key microbial species, gaps in specific microbial functions and, as a consequence, an impaired modulation of the gut wall activity. This may lead to the colonization of pathogenic microorganisms - causing diarrhea or necrotizing enteritis (Sekirov et al, 2008).
  • CDAD Crohn's disease
  • probiotics are mostly comprised of 1 to a couple of not interconnected microbial strains (mostly lactic acid producing bacteria) with a specific functionality.
  • FMT fecal microbial transplants
  • WO2014145958A2 Network-based microbial compositions and methods
  • the plurality of isolated bacteria or the purified bacterial preparation is able to form a so-called network ecology.
  • the bacteria belonging to this preparation are selected based on genomic information and are provided to the mammalian subject as a loosely assembled set of strains.
  • Van den Abbeele et al. (2013) suggested the possibility of creating a glycan-degrading community by using conventional in vitro fermenters which can be inoculated with relevant keystone species and a mixture of cross-feeding microbes. After inoculation and stabilization, such a microbial network unit for specific functions can be attained and produced at large scale.
  • the present invention relates in first instance to a composition consisting essentially of bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae preferably for use to prevent or treat symptoms associated with a gastro-intestinal disorder.
  • the present invention relates to a method to prevent or treat symptoms associated with a gastro-intestinal disorder in a subject in need thereof comprising administering a therapeutically effective amount of a composition consisting essentially of bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • the present invention further relates to a composition as described above wherein said gastro-intestinal disorder is a disruption of the barrier function of the gut, diarrhea, constipation, irritable bowel syndrome, inflammatory bowel disease, Crohn's disease, ulcerative colitis, coeliac disease, pouchitis, mucositis, an infection of the gut, gut microbiota dysbiosis or any combination thereof.
  • the present invention also relates to a composition as described above wherein said gastro-intestinal disorder is prevented or treated via: a) stimulating growth and/or activity of one or a limited number of beneficial bacteria in the intestinal tract, b) inhibiting growth and/or activity of one or a limited number of pathogenic bacteria in the intestinal tract, c) relatively increasing the attachment of non-pathogenic bacteria to the mucosa of the gastrointestinal surface, d) reducing uncontrolled uptake of antigens, pro-inflammatory, bacteria or bacterial products by the gut, e) providing anti-inflammatory activity at the intestinal surface, f) increasing gut barrier functioning, g) producing bacterial metabolites or h) any combination of a) to g).
  • the present invention also relates to a composition as described above wherein bacteria belonging to the species Roseburia hominis are eliminated from said composition.
  • the present invention further relates to a composition as described above wherein bacteria belonging to the species Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron and Bacteroides vulgatus are further added to said composition.
  • the present invention further relates to a composition as described above further comprising one or more prebiotics.
  • the present invention relates to a composition as described above wherein said bacteria are preadapted by growing them together in a fermenter prior to administering said composition to prevent or treat said gastro-intestinal disorders.
  • the present invention further relates to a composition as described above wherein said fermenter is a dynamic simulator of the gastro-intestinal tract.
  • the present invention relates to a composition as described above wherein said bacteria are chosen from the list of the following strains: Faecalibacterium prausnitzii LMG P-29362, Faecalibacterium prausnitzii DSMZ 17677, Butyricicoccus pullicaecorum LMG P-29360, Butyricicoccus pullicaecorum LMG24109, Roseburia inulinivorans LMG P-29365, Roseburia inulinivorans DSMZ 16841, Roseburia hominis LMG P-29364, Roseburia hominis DSMZ 16839, Akkermansia muciniphila LMG P-29361, Akkermansia muciniphila DSMZ 22959, Lactobacillus plantarum LMG P-29366, Lactobacillus plantarum ZJ316, Anaerostipes caccae LMG P-29359, Anaerostipes caccae DSMZ 14662 and/or strain
  • the present invention further relates to a composition as described above wherein said composition is a pharmaceutical composition formulated either as a rectally administrated form or an orally ingestible form.
  • the present invention further relates to a composition as described above wherein said orally ingestible form is a capsule, microcapsule, tablet, granule, powder, troche, pill, suspension or syrup.
  • the present invention further relates to a composition as described above which is incorporated in a food, drink, food supplement or nutraceutical.
  • the present invention more specifically relates to a composition as described above wherein said composition comprises between 10 5 and 10 11 colony forming units of bacteria.
  • the gut microbiome comprises hundreds of microbial species that co-exist within different subjects and that interact with each other and the host.
  • the gut microbiota play a key role in human health and disease by regulating metabolic functions and immune homeostasis (Cout et al, 2014).
  • Several studies have investigated these complex gut microbial communities in an attempt to define a "core microbiome", implying that all human individuals share a key number of essential species or strains that define the functional capabilities of a healthy gut microbiome (Kinross et al., 2011).
  • the extensive literature that is available on the composition and function of the gut microbiota (eg. keystone species, mucosal versus luminal microbiota, proximal versus distal colon bacteria, etc.) and functional genome analysis, a list of microbial candidates could be identified that covers the main functionalities of the complex human gut microbiome.
  • the present invention relates in first instance to a specific selection of a subgroup of bacterial species of the human gut microbiome which have a particular and surprising effect. More specifically , the present invention relates to a composition consisting essentially of bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae preferably for use to prevent or treat symptoms associated with a gastro-intestinal disorder.
  • the term 'consisting essentially of' indicates that said composition may include other bacterial species and/or other components provided they do not negatively affect the effect (i.e.
  • a composition of the invention comprises bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • a composition of the invention consists of bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • the bacterial species Faecalibacterium prausnitzii (Duncan et al. 2002), Butyricicoccus pullicaecorum (Eeckhaut et al. 2008), Roseburia inulinivorans (Duncan et al. 2006), Roseburia hominis (Duncan et al. 2006), Akkermansia muciniphila (Derrien et al. 2004), Lactobacillus plantarum (Walter 2008) and Anaerostipes caccae (Schwiertz et al. 2002) are well known bacterial species to a skilled person.
  • the terms 'symptoms associated with a gastro-intestinal disorder' refer to health problems in humans and animals.
  • composition of the present invention leads more specifically to prevention/recovery from dysbiosis resulting in a positive modulation of the interaction between bacteria and intestinal surface.
  • an improved functioning of the intestinal surface is obtained: e.g. barrier, hormonal, immune functioning.
  • the onset of the effect on the intestinal surface is quicker when a 'pre-adapted composition' is dosed as compared to a loosely assembled set of the same strains' (see further).
  • modulating or improving the barrier, hormonal or immune function of the intestinal surface is meant to include altering any parameter that affects the normal homeostasis of the intestinal surface and in particular its role in the first line defense against the invasion by pathogens, antigens or other harmful substances and its role to produce substances (e.g. immune molecules, hormones) which have systemic influences on the host.
  • Said parameters include, but are not limited to:
  • the present invention thus relates to a composition as described above wherein said gastro-intestinal disorder is prevented or treated via: a) stimulating growth and/or activity of one or a limited number of beneficial bacteria in the intestinal tract, b) inhibiting growth and/or activity of one or a limited number of pathogenic bacteria in the intestinal tract, c) relatively increasing the attachment of non-pathogenic bacteria to the mucosa of the gastrointestinal surface, d) reducing uncontrolled uptake of antigens, pro-inflammatory, bacteria or bacterial products by the gut, e) providing anti-inflammatory activity at the intestinal surface, f) increasing gut barrier functioning, g) producing bacterial metabolites or h) any combination of a) to g).
  • Health conditions which may be associated with general gastro-intestinal disorders include -but are not limited to-: constipation, Irritable Bowel Syndrome (IBS), Inflammatory Bowel Diseases (IBD), gut microbiota dysbiosis, mucositis, metabolic syndrome, obesity, diabetes, a cardiovascular disease, chronic fatigue syndrome, a mental condition, impaired cognitive function, a neurodegenerative disease, a form of cancer, an autoimmune condition, impaired immune functioning, rheumatism, rheumatoid arthritis, inflammation of the female reproductive apparatus, infection of pathogens (bacteria, viruses and fungi).
  • neurodegenerative diseases include, but are not limited to ALS, dementia, Alzheimer's, Parkinson's and Huntington's disease.
  • Examples of types of cancers include, but are not limited to lung cancer, breast cancer, prostate cancer, pancreatic cancer and particularly colorectal cancer.
  • Examples of autoimmune diseases include, but are not limited to multiple sclerosis, atopic dermatitis, celiac disease, psoriasis and lupus.
  • compositions of the present invention enhance the interaction and/or activity of non-pathogenic bacteria to the mucosal layer of the gastrointestinal epithelium, it is envisaged that said preparations are particularly useful to improve the barrier function of the intestinal surface, such as for example to prevent or reduce the uncontrolled uptake from the gut of antigens, pro-inflammatory molecules, pathogenic bacteria or bacterial products.
  • One such indication, with an impaired mucosal barrier is Inflammatory Bowel Disease.
  • Inflammatory Bowel Diseases mucosal injury with an impaired resolution of the lesions is one of the key elements that lead to these chronic indications, the compositions of the present invention, have a beneficial effect in said indication.
  • compositions of the present invention in the prevention and treatment of conditions associated with an impaired barrier function and characterized by the uncontrolled uptake from the gut of antigens, pro-inflammatory molecules, pathogenic bacteria or bacterial products.
  • Inflammatory bowel diseases also referred to as “chronic colonic diseases”, as used herein include any condition characterized by persistent mucosal inflammation at different levels of the gastrointestinal tract, such as for example inflammatory bowel syndrome, mucositis, gastric ulcers, Crohn's disease, ulcerative colitis, colorectal cancer and pouchitis.
  • mucositis is generally recognized as being essentially characterized by inflammation of the mucosal surface lining the mouth and gastrointestinal tract, typically as adverse event of chemotherapy and radiotherapy or stem cell transplantation, it is also to be envisaged that the application of the compositions of the present invention have a beneficial effect in said indication. It is accordingly an objective of the present invention, to provide the use of the compositions of the present invention in the prevention and treatment of conditions associated with mucositis. Mucositis can occur anywhere along the gastrointestinal tract. In case of occurring in the oral cavity, it is typically referred to as oral mucositis.
  • compositions of the present invention provide protection against invasion by antigens that cause allergic reactions, whereby such allergens may comprise certain food substances, chemicals and other molecules.
  • allergens may comprise certain food substances, chemicals and other molecules.
  • the present invention provides the use of compositions in the prevention and treatment of conditions associated with the invasion by antigens that cause allergic reactions (eg food allergies, asthma, eczema)
  • compositions do influence both the gut-associated lymphoid tissue (GALT) as well as the systemic immune system. Among other effects this may result in decreased expression of pro-inflammatory cytokines and increased production of immunoregulatory factors and improved activity of lymphocytes. It is therefore envisaged that said compositions are particularly useful in improving the development and functioning of the host immune system.
  • GALT gut-associated lymphoid tissue
  • compositions of the present invention modulate the epithelial barrier and subsequently decrease chronic inflammation
  • said compositions are particularly useful in controlling and improving metabolic homeostasis.
  • Non-limiting effects of said preparations on metabolic homeostasis include control of food intake and fat and glucose metabolism, improvement of insulin secretion and sensitivity and control of cholesterol synthesis and metabolism.
  • It is accordingly an objective of the present invention to provide the use of the compositions of the present invention in the management of food uptake, induction of satiety, weight management, the prevention and treatment of conditions associated with an impaired metabolic homeostasis, such as obesity and type 2 diabetes.
  • compositions of the present invention decrease several established causal risk factors of cardiovascular diseases (CVD), it is to be envisaged in another aspect of the invention that said compositions are particularly useful for the prevention of CVD.
  • CVD technically refers to any disease that affects the cardiovascular system, yet is usually used to refer to those related to atherosclerosis.
  • the latter is a syndrome affecting arterial blood vessels, a chronic inflammatory response in the walls of arteries, in large part due to the accumulation of macrophage white blood cells and promoted by low density lipoproteins.
  • CVD development depends on multiple mechanisms and a number of clear causal risk factors have been identified.
  • LDL-ox LDL becoming oxidized (LDL-ox) by free radicals, particularly oxygen free radicals, in situations of oxidative stress.
  • LDL-ox oxidized oxidized
  • Excessive response of the immune system, in case of chronic inflammation, to damage caused by LDL-ox further promotes the expansion of the disease. It is accordingly an objective of the present invention, to provide the use of the compositions of the present invention in the prevention or treatment of CVD.
  • compositions of the present invention provides protection against mucosal attachment and invasion by pathogens.
  • pathogens include, but are not limited to Bacillus anthracis; Bacillus cereus; Bordetella pertussis; Borrelia burgdorferi; Brucella abortus; Brucella canis; Brucella melitensis; Brucella suis; Campylobacter jejuni; Chlamydia pneumonia; Chlamydia trachomatis; Chlamydophila psittaci; Clostridium botulinum; Clostridium difficile; Clostridium perfringens; Clostridium tetani; Corynebacterium diphtheria; Enterotoxigenic Escherichia coli (ETEC); Enteropathogenic E.
  • pathogens include, but are not limited to Bacillus anthracis; Bacillus cereus; Bordetella pertussis; Borrelia burgdorferi; Brucella abortus
  • the present invention thus relates to a method to prevent or treat symptoms associated with a gastro-intestinal disorder in a subject in need thereof comprising administering a therapeutically effective amount of a composition consisting essentially of bacteria belonging to the species Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • the term 'subject in need' refers a human or a non-human animal having a gastro-intestinal disorder as described above.
  • a therapeutically effective amount' refers to a minimum of the combined total amount of the 7 bacterial species that is capable to exert its prophylactic or therapeutic effect.
  • the 7 bacteria species are listed hereafter: Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae
  • a therapeutically effective amount may also refer to a minimum of a combined total amount of 6 bacteria species listed hereafter: Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • said combined total amount can be the result of equal amounts of each of the 7 bacterial species or unequal amounts of the 7 bacterial species, in which each single species of the 7 bacterial species has a minimum abundance of 0.0001% of the combined total amount, more preferably a minimum abundance of 0.001% of the combined total amount and most preferably a minimum abundance of 0.01% of the combined total amount. If thus -for example- 6 species have an abundance of 10.00% of the combined total amount, then the 7 th species has an abundance of 40.00 % of the combined total amount.
  • said combined total amount ranges between a daily dose of 10 2 and 10 14 bacterial cells, preferably ranges between a daily dose of 10 3 and 10 13 bacterial cells, more preferably ranges between a daily dose of 10 4 and 10 12 bacterial cells and most preferably ranges between a daily dose of 10 5 and 10 11 bacterial cells.
  • the present invention further relates to a composition as described above wherein bacteria belonging to the species Roseburia hominis are eliminated from said composition.
  • the term 'eliminated' refers in particular to making a composition of 6 bacterial species as is indicated further in the Examples section without adding or removing the species Roseburia hominis as a 7 th species.
  • the present invention further relates to a composition as described above wherein bacteria belonging to the species Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron and Bacteroides vulgatus are further added to said composition.
  • the bacterial species Escherichia coli (Rath et al. 1999), Enterococcus faecium (Schleifer et al. 1984), Lactobacillus mucosae (Roos et al. 2000), Bifidobacterium adolescentis (Scharek et al. 2000), Bifidobacterium longum (Bahaka et al. 1993), Bacteroides thetaiotaomicron (Scharek et al. 2000) and Bacteroides vulgatus (Rath et al. 1999) are well known bacterial species to a skilled person.
  • the present invention further relates to a composition as described above further comprising one or more prebiotics.
  • prebiotic' refers to any chemical that induce the growth or activity of microorganisms (e.g., bacteria) that contribute to the well-being of their host.
  • microorganisms e.g., bacteria
  • prebiotics can influence or alter the composition of organisms in the gut microbiome.
  • Typical -but non-limiting- prebiotics are non-digestible fiber compounds that at least partially pass undigested through the upper part of the gastrointestinal tract and stimulate the growth or activity of advantageous bacteria that colonize the large bowel by acting as substrate for them.
  • the present invention relates to a composition as described above wherein said bacteria are grown together in a fermenter prior to administering said composition to prevent or treat said gastro-intestinal disorders.
  • the latter compositions are also referred to (see further) as the "Collaborome strategy” or as the “Alternative collaborome strategy”.
  • composition wherein said bacteria are not grown together in a fermenter prior to administration are referred to (see further) as the "Assembly strategy".
  • the present invention further relates to a composition as described above wherein said fermenter is a dynamic simulator of the gastro-intestinal tract.
  • said fermenter is a dynamic simulator of the gastro-intestinal tract.
  • the latter compositions are also referred to (see further) as the "Collaborome strategy”.
  • the SHIME® (Simulator of the Human Microbial Ecosystem) is a dynamic in vitro model of the human gastrointestinal tract that is composed of five double-jacketed vessels, simulating the stomach, small intestine, and the three colon regions (ascending, transverse, and descending colon), with a total retention time of 72 h ( Fig. 1 ). Three times per day, 140 ml SHIME® feed and 60 ml pancreatic juice were added to the stomach and small intestine compartments, respectively (Van den Abbeele et al., 2010). After an initial 2-week stabilization period - which allows the microbiota to adapt to the imposed in vitro conditions - the isolation procedure was started.
  • the selected microbial strains of the present invention can thus be inoculated in single stage (alternative collaborome strategy) or multi-stage reactors or dynamic simulators of the gastrointestinal tract (e.g. SHIME® or M-SHIME®, collaborome strategy) under standardized conditions representative for the GI tract.
  • a reactor comprising a composition comprising, consisting of or consisting essentially of bacteria belonging to 6 or 7 or up to 14 species as defined herein and further listed below:
  • this reactor comprising said composition is under standardized conditions representative for the GI tract as defined below.
  • the parameters characterizing the standardized conditions include but are not limited to: pH (range between 1.5 and 8); availability of carbon sources (either carbohydrate or proteins or a combination thereof); retention time in a specific reactor (range between 10 min and 200 h); oxygen availability (range between 0 and 8 g/L); availability of micronutrients; presence/absence of antibiotics; concentration of bile salts (range between 0 and 20 mM); presence of heavy metals; presence of host factors as immune molecules.
  • the parameters characterizing the standardized conditions comprise pH, retention time in a specific reactor and concentration of bile salts, all as earlier defined herein. Depending on the complexity of the collaborome, a period of 1 to 15 days is needed to obtain a functionally stable collaborome.
  • a composition as defined herein is therefore obtainable after having been trained or cultured during a time between 3 and 10 days under conditions wherein pH, retention time in a specific reactor and concentration of bile salts have been set as defined herein. Such a process allows the production of a composition or collaborome which is functionally stable.
  • a functionally stable collaborome is a composition as defined herein still comprising the initial different number of species of bacteria after at least 3 or 5 or 10 days of culture.
  • a reactor operating under standardized conditions representative for the GI tract, comprising: pH range between 1.5 and 8; availability of carbon sources; retention time between 10 min and 200 h; oxygen availability between 0 and 8 g/L; availability of micronutrients; presence/absence of antibiotics; concentration of bile salts between 0 and 20 mM; presence of heavy metals; presence of host factors as immune molecules.
  • said reactor is such that the parameters characterizing the standardized conditions comprise pH, retention time in a specific reactor and concentration of bile salts as defined in the previous paragraph.
  • such reactor comprises a composition of 5 and 20 distinct bacteria members, or 6 to 14 distinct bacteria members or 5 to 15 distinct bacteria members.
  • such composition resides for a time between 3 and 14 days or 3 and 10 days in such a reactor to obtain a functionally stable collaborome.
  • a set of microorganisms pre-adapted to function together takes the name of the 'collaborome strategy' or alternative collaborome strategy'.
  • the invention relates to pre-adapted compositions of sets of microbial strains preferably for use to significantly decrease the time of biotherapeutic onset and/or to significantly increase the effect of treatment of dysbiosis as compared to a loosely assembled set of the same microbial strains.
  • the efficacy depends on the endpoint for which the set of microorganisms has been designed. Possible functionalities include but are not limited to Short Chain Fatty Acid (SCFA) production; improvement in gut barrier permeability; decrease/increase in pro-inflammatory cytokines; increase in anti-inflammatory cytokines; decrease in pathogen concentration (at least 0.5 log); decrease in gas production; stimulation of specific gut-wall receptors; etc... Any value below 5% is considered physiologically not relevant.
  • SCFA Short Chain Fatty Acid
  • the present invention more specifically relates to a method to prevent or treat dysbiosis of humans and animals in need thereof comprising administering a therapeutic amount of a pre-adapted composition of a set of microbial strains to said humans or animals wherein said treatment results in a faster biotherapeutic onset and/or increased efficiency as compared to the administration of a loosely assembled set of the same microbial strains.
  • the present invention relates to a composition as described above wherein said bacteria are chosen from the list of the following strains: Faecalibacterium prausnitzii LMG P-29362, Faecalibacterium prausnitzii DSMZ 17677, Butyricicoccus pullicaecorum LMG P-29360, Butyricicoccus pullicaecorum LMG24109, Roseburia inulinivorans LMG P-29365, Roseburia inulinivorans DSMZ 16841, Roseburia hominis LMG P-29364, Roseburia hominis DSMZ 16839, Akkermansia muciniphila LMG P-29361, Akkermansia muciniphila DSMZ 22959, Lactobacillus plantarum LMG P-29366, Lactobacillus plantarum ZJ316, Anaerostipes caccae LMG P-29359, Anaerostipes caccae DSMZ 14662 and/or strain
  • 16S rRNA refers to a nucleic acid sequence of about 1542 nucleotides which is a component of the small prokaryotic ribosomal subunit (30S).
  • the 16S rRNA is known to act as a scaffold defining the positions of the ribosomal proteins.
  • the 16S rRNA sequence is commonly used for phylogenetic studies, as it is known to be a highly conserved sequence. Comparative analysis of 16S rRNA sequences from thousands of organisms has demonstrated the presence of oligonucleotide signature sequences.
  • the term "homology" refers to the sequence similarity of the nucleic acids. For example, in general, if two nucleic acids have identical sequences they show 100% homology. A change in the nucleotide sequence of one of the nucleic acids reduces the percentage of homology. In general, the percentage homology quantifies the degree of identity between two nucleic acid sentences.
  • Sequence identity or sequence homology is herein defined as a relationship between two or more amino acid (polypeptide or protein) sequences or two or more nucleic acid (polynucleotide) sequences, as determined by comparing the sequences. Usually, sequence identities or similarities are compared over the whole length of the sequences compared. In the art, “identity” also means the degree of sequence relatedness between amino acid or nucleic acid sequences, as the case may be, as determined by the match between strings of such sequences. "Similarity" between two amino acid sequences is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. "Identity” and “similarity” can be readily calculated by various methods, known to those skilled in the art. In a preferred embodiment, sequence identity is determined by comparing the whole length of the sequences as identified herein.
  • Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Preferred computer program methods to determine identity and similarity between two sequences include e.g. the BestFit, BLASTP, BLASTN, and FASTA ( Altschul, S. F. et al., J. Mol. Biol. 215:403-410 (1990 ), publicly available from NCBI and other sources ( BLAST Manual, Altschul, S., et al., NCBI NLM NIH Bethesda, MD 20894 ). A most preferred algorithm used is EMBOSS ( http://www.ebi.ac.uk/emboss/align ).
  • Preferred parameters for amino acid sequences comparison using EMBOSS are gap open 10.0, gap extend 0.5, Blosum 62 matrix.
  • Preferred parameters for nucleic acid sequences comparison using EMBOSS are gap open 10.0, gap extend 0.5, DNA full matrix (DNA identity matrix).
  • amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulphur-containing side chains is cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, and asparagine-glutamine.
  • Substitutional variants of the amino acid sequence disclosed herein are those in which at least one residue in the disclosed sequences has been removed and a different residue inserted in its place.
  • the amino acid change is conservative.
  • Preferred conservative substitutions for each of the naturally occurring amino acids are as follows: Ala to ser; Arg to lys; Asn to gln or his; Asp to glu; Cys to ser or ala; Gln to asn; Glu to asp; Gly to pro; His to asn or gin; Ile to leu or val; Leu to ile or val; Lys to arg; gln or glu; Met to leu or ile; Phe to met, leu or tyr; Ser to thr; Thr to ser; Trp to tyr; Tyr to trp or phe; and, Val to ile or leu.
  • 16s rRNA sequences can be deposited online, for example at GenBank ( http://www.ncbi.nlm.nih.gov/genbank/ ) and that they can be retrieved based on their unique accession number for use as reference 16S rRNA sequence in evaluation of sequence homology, as for example described by Eeckhaut et al. (2008).
  • GenBank accession numbers for the 16S rRNA sequences of 7 bacterial species of the composition are listed below. These accession numbers can be used to retrieve the respective 16S rRNA sequences from http://www.ncbi.nlm.nih.gov/genbank/ for assessment of sequence homology.
  • the present invention further relates to a composition as described above wherein said composition is a pharmaceutical composition formulated either as a rectally administrated form or an orally ingestible form.
  • the present invention further relates to a composition as described above wherein said orally ingestible form is a capsule, microcapsule, tablet, granule, powder, troche, pill, suspension or syrup.
  • the present invention further relates to a composition as described above which is incorporated in a food, drink, food supplement or nutraceutical.
  • the present invention thus relates to a composition as described above which is used as food, food supplement or medicine for a human, a non-human domestic or farmed land animal or an aquatic animal.
  • the composition can thus be introduced in food, functional foods, food supplements, cosmetics, nutraceutical formulations, probiotic composition or pharmaceutical.
  • a food is typically an edible material composed primarily of one or more of the macronutrients protein, carbohydrate and fat.
  • a food may also contain one or more micronutrients such as vitamins or minerals.
  • the term food as used herein also covers a beverage.
  • foods in which the composition may be incorporated include snack bars, cereals, buns, muffins, biscuits, cakes, pastries, processed vegetables, sweets, probiotic formulations including yoghurts, beverages, plant oil-based liquids, animal fat-based liquids, frozen confections and cheeses.
  • Preferred foods include yoghurts, cheeses and other dairy products.
  • beverages include soft beverages, syrups, squashes, dry drink mixes and nutritional beverages.
  • a nutraceutical is a food ingredient, food supplement or food product which is considered to provide a medical or health benefit, including the prevention and treatment of disease.
  • a functional food is a food that is typically marketed as providing a health benefit beyond that of supplying pure nutrition to the consumer.
  • the invention also provides a probiotic comprising a composition discussed herein.
  • a probiotic is typically a live supplement which can enhance the intestinal microbiota. Such probiotics may be given in particularly to humans but also to farm and domestic animals and to aquatic organisms.
  • the probiotic may additionally comprise one or more acceptable excipients or flavorings, which are suitable for ingestion by a human or animal.
  • composition of the invention may be used in the production of pharmaceutical compositions.
  • the invention further provides a pharmaceutical composition comprising a composition of the invention and a pharmaceutically acceptable excipient or carrier.
  • compositions comprising compounds of the invention may be in diverse forms, for example in the form of a tablet, capsule or powder.
  • excipients which may be present in such compositions include diluents (e.g. starch, cellulose derivatives or sugar derivatives), a stabilizer (e.g. hygroscopic excipients such as silica or maltodextrin), a lubricant (e.g. magnesium stearate), a buffer (e.g. phosphate buffer), a binder, coating, preservative or suspension agent.
  • diluents e.g. starch, cellulose derivatives or sugar derivatives
  • a stabilizer e.g. hygroscopic excipients such as silica or maltodextrin
  • a lubricant e.g. magnesium stearate
  • buffer e.g. phosphate buffer
  • a binder coating, preservative or suspension agent.
  • the present invention more specifically relates to a composition as described above wherein said composition comprises a total between 10 5 and 10 11 colony forming units of bacteria of the present invention.
  • the verb "to comprise” and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded.
  • the verb "to consist” may be replaced by "to consist essentially of” meaning that a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
  • reference to an element by the indefinite article “a” or “an” does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements.
  • the indefinite article “a” or “an” thus usually means “at least one”.
  • a young, healthy donor with no prior exposure to antibiotic therapy was selected to inoculate the SHIME® model.
  • the SHIME® set up was used for isolation of bacterial strains with different functional properties, such as fiber degraders (e.g. Bifidobacteria, Bacteroides), fermentative (e.g. Escherichia coli ) or lactate producers (e.g.
  • Lactobacilli Lactobacilli, Pediococci and Enterococci
  • butyrate producers e.g. Anaerostipes caccae, Butyricicoccus pullicaecorum, Faecalibacteruim prausnitzii, Roseburia hominis, Roseburia inulinivorans, Clostridium butyricum
  • propionate producers e.g. Bacteroides thetaiotaomicron, Bacteroides vulgatus, Roseburia inulinivorans, Akkermansia muciniphila).
  • selective media were selected such as LAMVAB (lactobacilli; Hartemink et al 1997), RB (bifidobacteria; Hartemink et al. 1996), Enterococcus medium (Enterococci; Possemiers et al. 2004), TBX ( Escherichia coli; Le Bon et al. 2010), BBE ( Bacteroides fragilis group; Livingston et al. 1978), Mucin minimal medium (Akkermansia; Derrien et al. 2004), M2GSC (butyrate producers; Barcenilla et al.
  • LAMVAB lactobacilli; Hartemink et al 1997)
  • RB bifidobacteria; Hartemink et al. 1996)
  • Enterococcus medium Enterococci; Possemiers et al. 2004
  • TBX Escherichia coli; Le Bon et al. 2010
  • BBE Bacteroides fragilis
  • pure cultures isolated from the SHIME® reactor and fecal were used (as described in example 1.1). Additionally, pure cultures were sourced from culture collections such as BCCM/LMG ( http://bccm.belspo.be ) and DSMZ ( www.dsmz.de ).
  • Short-chain fatty acids are the end products of dietary fibers fermentation by the intestinal microbiota and are known to exert several beneficial effects on host health.
  • the main SCFA produced are acetate, butyrate and propionate in an approximate 60:20:20 molar ratio.
  • acetate can be absorbed from the gut and used as energy substrate by the host, butyrate acts as main energy source for the gut epithelium and has proven protective effects against inflammation and colon cancer.
  • Propionate has similar local activity in the gut as compared to butyrate, yet it is also transported to the liver where it was shown to have positive cholesterol-lowering effects and effects on glycemic control.
  • Isolate-X a total 20 isolates with the most interesting fermentation profiles, obtained from the isolation and selection round as described in 1.1 (referred to as "Isolate-X") or ordered from culture collections, were retrieved from their glycerol stocks and grown under their respective optimal growth conditions to obtain homogeneous suspensions of the bacterial strains.
  • Ref. Species Strain Ref. Species Strain 1 Lactobacillus plantarum Isolate-1 11 Faecalibacterium prausnitzii Isolate-11 2 Clostridium bolteae Isolate-2 12 Roseburia inulinivorans Isolate-12 3 Desulfovibrio desulfuricans Isolate-3 13 Ruminococcus spp.
  • Isolate-13 4 Akkermansia muciniphila Isolate-4 14 Lacobacillus acidophilus Isolate-14 5 Coprococcus spp. Isolate-5 15 Enterococcus faecium Isolate-15 6 Roseburia hominis Isolate-6 16 Butyrivibrio fibrisolvens Isolate-16 7 Bacteroides thetaiotaomicron Isolate-7 17 Eubacterium limosum DSM20543 8 Clostridium butyricum Isolate-8 18 Escherichia coli Nissle 1917 9 Anaerostipes caccae Isolate-9 19 Eubacterium rectale DSM17629 10 Bifidobacterium adolescentis Isolate-10 20 Butyricicoccus pullicaecorum Isolate-17
  • Isolates were combined in numbers ranging from 2 to 10 in a set of 98 individual initial screening experiments. For each experiment, fermentation was started in sterile incubation bottles containing sterilized SHIME® nutritional medium adjusted to pH 6.8 with KH 2 PO 4 /K 2 HPO 4 and flushed with nitrogen. Then the sterilized medium was inoculated with 10% (v/v) of mixed inoculum consisting of equal volumes of the selected species. Incubation bottles were flushed with nitrogen to ensure anaerobic conditions and were incubated at 37°C (90 rpm). Samples were analyzed after 24h for SCFA production.
  • Fig. 2 describes butyrate levels obtained upon 24h incubation of the 23 different compositions as well as their effect on the TEER values. Strong variation was observed in both butyrate levels and effects on gut barrier functioning and combinations with highest butyrate levels not necessarily induced highest protective effects on TEER levels, as shown by different ranking.
  • M7-B in Fig. 2 one composition of 7 different isolates was ranked first on both butyrate levels after 24h and especially on protective effects towards gut barrier function.
  • This composition contained 6 isolates from the SHIME and one culture obtained from a human fecal sample.
  • M7-B was composed of novel SHIME isolates of Lactobacillus plantarum, Faecalibacterium prausnitzii, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila and Anaerostipes caccae and of a novel fecal isolate of Butyricicoccus pullicaecorum.
  • the novel isolates were all present in at least one of the other compositions shown in Fig. 2 , yet none of the other compositions reached the same effectivity with respect to butyrate production and protection of TEER values. This shows that the observed effect is not related to one of the specific species present in the composition, but that only the specific combination of these 7 bacteria leads to the surprising positive results.
  • the 7 novel isolates were deposited at the BCCM/LMG Bacteria collection (Ghent Belgium), with accession numbers: Faecalibacterium prausnitzii LMG P-29362, Butyricicoccus pullicaecorum LMG P-29360, Roseburia inulinivorans LMG P-29365, Roseburia hominis LMG P-29364, Akkermansia muciniphila LMG P-29361, Lactobacillus plantarum LMG P-29366 and Anaerostipes caccae LMG P-29359.
  • compositions excluding Roseburia hominis still resulted in a similar functionality of the complete composition after 48h of incubation, one can also envisage -as second best- the use of the composition of 6 species, consisting essentially of Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Akkermansia muciniphila, Lactobacillus plantarum and Anaerostipes caccae.
  • a composition consisting of the species Lactobacillus plantarum, Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila and Anaerostipes caccae is produced using 3 different strategies. These strategies include either 1) growing the species of the composition separately, followed by mixing them together, 2) growing the species of the composition together in a multi-stage fermenter (i.e. the in vitro SHIME® model as described above) and 3) growing the species of the composition together in a single-stage fermenter.
  • the selected species were retrieved from their glycerol stocks and grown under their respective optimal growth conditions to obtain homogeneous suspensions of the bacterial strains.
  • a mixed inoculum was created consisting of equal volumes of the selected species. This inoculum was added at 10% (v/v) to sterile incubation bottles containing sterilized SHIME® nutritional medium adjusted to pH 6.8 with KH 2 PO 4 /K 2 HPO 4 . Incubation bottles were flushed with nitrogen to ensure anaerobic conditions and were incubated at 37°C (90 rpm).
  • Conditioned SHIME® nutritional medium was prepared by incubating 700 mL of normal SHIME® feed (pH2) for 1 hour at 37°C, after which 300 mL of pancreatic juice (pH 6.8) - supplemented with 25g/L NaHCO3, 23.6 g/L KH 2 PO 4 and 4.7 g/L K 2 HPO 4 - was added. Samples were analyzed over a period of 5 days for SCFA production ( Fig. 4 ). Butyrate levels reached 7 mM upon 24h incubation of the assembly and a maximum of 14 mM after 5 days.
  • the selected species were retrieved from their glycerol stocks and grown under their respective optimal growth conditions to obtain homogeneous suspensions of the bacterial strains. Then, the strains were mixed and inoculated in triplicate in a SHIME® setup (Van den Abbeele et al., 2010) consisting of a single colon region at a pH of 6.15-6.4. A two-week adaptation period was implemented to create a functional collaborome composition. The need and relevance of such an adaptation period is clearly demonstrated by the evolution of SCFA profiles during the cultivation of the composition of selected species ( Fig. 5 ).
  • the composition requires time to adapt to one another and to become active in converting the supplied substrates to SCFA.
  • four to six days after inoculation the production of SCFA by the composition started to stabilize and high levels of butyrate were measured.
  • day 14 the final day of incubation
  • each of the triplicate incubations resulted in a highly-similar, stable an strongly active functional composition with butyrate levels reaching 19 mM.
  • the production of said composition was undertaken using an optimized single stage fermenter approach, operated in fed-batch mode (i.e. the alternative "Collaborome” strategy or the strategy "wherein said bacteria are grown together in 'a' fermenter prior to administration”).
  • the selected species were retrieved from their glycerol stocks and grown under their respective optimal growth conditions to obtain homogeneous suspensions of the bacterial strains. Fermentation was started in sterile incubation bottles containing sterilized SHIME® feed adjusted to pH 6.8 with KH 2 PO 4 /K 2 HPO 4 and flushed with nitrogen. Then the sterilized medium was inoculated with 10% (v/v) of mixed inoculum consisting of equal volumes of the selected species.
  • Incubation bottles were flushed with nitrogen to ensure anaerobic conditions and were incubated at 37°C (90 rpm). At specific intervals of 16h, 40% (v:v) of the growth medium was replaced with conditioned SHIME® nutritional medium.
  • Conditioned SHIME® nutritional medium was prepared by incubating 700 mL of normal SHIME® feed (pH2) for 1 hour at 37°C, after which 300 mL of pancreatic juice (pH 6.8) - supplemented with 25g/L NaHCO3, 23.6 g/L KH 2 PO 4 and 4.7 g/L K 2 HPO 4 - was added.
  • the total SCFA production and the ratio of SCFA produced by the composition was stable after 6 replacement cycles.
  • the stabilized Collaborome led to a maximized SCFA production (acetate/propionate/butyrate ratio was around 14/12/74) 2 days earlier as compared to the same set of species in the assembly strategy and a 25% higher butyrate production.
  • a dysbiosed microbial composition is more susceptible to infections by pathogens.
  • a number of gastrointestinal diseases have been correlated with a dysbiosed microbial composition, such as inflammatory bowel diseases, underlining the importance of a healthy gut microbiome.
  • Recovery of the taxonomic composition and especially functionality after long-term antibiotic intake usually takes three months to reach the pre-treatment state, a healthy gut microbial community (Panda et al., 2014).
  • a decrease in the recovery time after exposure to antibiotic therapy could thus reduce the risk of severe infections and promote host health in general.
  • the observed functional activity of the selected composition could be a promising strategy to enhance restoration of microbial communities upon antibiotics-induced dysbiosis and reduce infection risks.
  • antibiotics-induced dysbiosis was modeled in the in vitro SHIME® model by dosing the appropriate antibiotics.
  • the aim of this experiment was to evaluate the recovery of the typical 'healthy' metabolite profiles in the simulated intestinal colon environments upon administration of the functional composition.
  • the experiment aimed to differentiate the effectivity of the composition, when either produced through the "Assembly” strategy or the "Collaborome” strategy (see example 1.3).
  • the composition was in this specific experiment further supplemented with Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron and Bacteroides vulgatus.
  • SHIME® vessels (pH 6.15-6.40) were inoculated with fecal material and allowed to stabilize during 14 days (M-SHIME® set up - Van den Abbeele et al., 2012).
  • M-SHIME® set up - Van den Abbeele et al., 2012 After a control period of 2 weeks, the SHIME®-derived colon microbiota was treated with a cocktail of antibiotics (40/40/10 mg/L of amoxicillin/ciprofloxacin/tetracyclin, respectively) to induce dysbiosis.
  • the dysbiosed microbiota was treated for 5 days with the functional composition, produced either through the "Assembly” strategy or the "Collaborome” strategy.
  • Endpoint of the study was to evaluate the recovery of the typical 'healthy' SCFA metabolite profiles in the simulated intestinal colon environments.
  • a control SHIME® vessel was included to simulate spontaneous recovery of the metabolic activity of the gut community after antibiotic exposure, without administration of the composition.
  • the results are expressed as the delta of SCFA levels in the SHIME at the each time point vs. the values before antibiotic administration ( Fig. 8 ).
  • composition of the Assembly strategy induced full recovery of propionate and butyrate after 5 days and 3 days, respectively
  • composition of the Collaborome strategy induced a faster recovery as opposed to the Assembly strategy with full recovery of propionate and butyrate after 4 days and 2.5 days, respectively.
  • Collaborome strategy also resulted in an increased final activity with increased propionate and butyrate levels as opposed to the Assembly strategy.
  • IBD Inflammatory Bowel Diseases
  • a state of gut microbiota dysbiosis includes a lower abundance of butyryl CoA:acetate CoA transferase and propionate kinase ( Vermeiren et al., FEMS 2011 ), which in turn negatively affects the production of a balanced SCFA production capacity.
  • SCFA butyryl CoA:acetate CoA transferase and propionate kinase
  • IBD associated dysbiosis was modeled in the in vitro M-SHIME® model, as described before (Vigsnaes et al. 2013).
  • the aim of this experiment was to evaluate the recovery of the microbiota in terms of SCFA profiles in the simulated intestinal colon environment upon administration of the functional composition.
  • the experiment aimed to differentiate the effectivity of the composition, when either produced through the "Assembly” strategy or the "Collaborome” strategy (see example 1.3).
  • SHIME® vessels (pH 6.15-6.40) were inoculated with fecal material from an Ulcerative Colitis patient (M-SHIME® set up - Van den Abbeele et al., 2012).
  • M-SHIME® Ulcerative Colitis patient
  • a single dose of the functional composition produced either through the "Assembly” strategy or the "Collaborome” strategy, was added to the colon region to simulate administration.
  • a third experiment ran in parallel as control experiment without administration of the composition. Production of acetate, propionate and butyrate was followed 1 and 2 days after administration of the composition.
  • a Clostridium difficile challenge test was performed aiming to evaluate whether the functional composition is not only functionally active under intestinal conditions, yet can also protect the intestinal environment against infections.
  • the composition is challenged with vegetative Clostridium difficile ( Cdif ) cells to assess its capacity to inhibit growth of Cdif under simulated gastro-intestinal conditions.
  • Cdif vegetative Clostridium difficile
  • the experiment aimed to differentiate the effectivity of the composition, when either produced through the "Assembly” strategy or the "Collaborome” strategy (see example 1.3).
  • a glycerol stock of Clostridium difficile (LMG 21717 T ) was thawed and inoculated in a bottle containing Reinforced Clostridial Medium (RCM) broth that was flushed with nitrogen to ensure anaerobic conditions.
  • the bottle was incubated in a shaking incubator (90 rpm) for 24 hours and 10% of the grown culture was again inoculated in RCM broth.
  • the homogenized C. difficile culture was aliquoted (in triplicate) in bottles (10% v:v) containing:
  • Bottles were incubated at 37°C in a shaking incubator (90 rpm). At regular time points, a sample was collected and immediately frozen at -80°C before quantifying C. difficile by means of a qPCR assay based on the detection and quantification of the triose phosphate isomerase gene.
  • genomic DNA was extracted according to Boon et al. (2003).
  • the amplification reaction included forward and reverse oligonucleotide: 5'- TATGGACTATGTTGTAATAGGAC-3' (forward) and 5'-CATAATATTGGGTCTATTCCTAC-3' (reverse). Absolute quantification of the PCR product was obtained by creating a standard curve.
  • compositions are functionally active under complex intestinal conditions and can restore intestinal metabolite profiles, with highest activity in case of the production through the Collaborome strategy. This may in turn beneficially influence the intestinal epithelium and thereby gut barrier functioning and local immunity.
  • this example describes the combination of samples collected from the previous experiments on an established co-culture cell model of enterocytes (Caco-2 cells) and macrophages (THP1) (Possemiers et al. 2013).
  • THP1 cells enterocytes
  • THP1 cells macrophages
  • stimulation of THP1 cells with LPS results in increased production of pro-inflammatory cytokines, which in turn tends to disrupt the enterocyte layer creating a so-called leaky gut' condition.
  • the effect on the leaky gut' is measured by assessing the effect of the transepithelial electrical resistance (TEER) [measurement for gut barrier efficiency] and inflammatory cytokine production, as compared to a control condition.
  • TEER transepithelial electrical resistance
  • composition 1 contains the specific isolates described in example 1.2
  • composition 2 is composed of strains from the same species obtained from culture collections:
  • the selected species were retrieved from their glycerol stocks and grown under their respective optimal growth conditions to obtain homogeneous suspensions of the bacterial strains. Then, the strains were mixed into Composition 1 and Composition 2, respectively, and each inoculated in triplicate in a SHIME® setup (Van den Abbeele et al., 2010) consisting of a single colon region at a pH of 6.15-6.4. Butyrate production profiles were followed up for a period of 14d.
  • the goal of the experiment in this example was to assess whether the functional composition can also in an in vivo setting restore the metabolic capacity of the gut microbiome after antibiotic-induced dysbiosis.
  • composition containing Lactobacillus plantarum, Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila and Anaerostipes caccae, was used and produced via the "Collaborome" strategy of Example 1.3.
  • composition was further supplemented with Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron and Bacteroides vulgatus (referred to as "extended composition").
  • compositions and “extended composition” were prepared fresh according to the Collaborome strategy, washed twice in PBS (in an anaerobic chamber to ensure anaerobic conditions), concentrated in 100 ⁇ L and administered to the mice via oral gavage as soon as possible.
  • Mice C57/BL6 of at least 5 weeks old were purchased, kept under pathogen-free conditions and fed a standard diet. Mouse experiments were performed in accordance with protocols approved by the Ethics Committee of Animal Trials of Ghent University, Belgium.
  • the antibiotic clindamycin was dosed to the drinking water at a concentration of 250 mg/L. After 5 days of antibiotic treatment, the stomach content of the mice was neutralized with NaHCO 3 after which the mice (10 mice per group) are orally gavaged for 5 consecutive days with:
  • a conventional group (without antibiotic treatment but treated with saline solution) is included as control to exclude variability arising from the gavage procedure.
  • fecal samples (approx. 100 mg/mouse) were collected and stored at -80°C for future analyses.
  • TNBS 2,4,6-trinitrobenzenesulfonic acid
  • the TNBS model is a commonly used model for colitis that mimics some of the features of Crohn's disease (Scheiffele et al. 2001), including weight loss, bloody diarrhea and intestinal wall thickening.
  • TNBS causes patchy transmural inflammation of the gut with the formation of deep ulcers, classical features found in patients with CD. This makes the TNBS model a good candidate for in vivo evaluation of the capacity of the functional composition to prevent and/or restore damage to the intestinal mucosa in IBD and to assist in maintaining/developing a healthy gut barrier.
  • the composition containing Lactobacillus plantarum, Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila and Anaerostipes caccae was used to evaluate the beneficial effects upon evaluation in the TNBS model. Furthermore, the experiment aimed to differentiate the effectivity of the composition, when either produced through the "Assembly” strategy or the "Collaborome” strategy (see example 1.3). Colitis was evoked in the animals by rectal instillation of TNBS, a mucosal sensitizing agents diluted in ethanol.
  • TNBS haptenizes the localized colonic and gut microbial proteins to become immunogenic, thereby triggering the host innate and adaptive immune responses.
  • a conventional group (without TNBS treatment but treated with saline solution) is included as control to exclude variability arising from the gavage procedure.
  • Disease Activity was monitored daily (before the daily treatment) by measuring body weight, fecal blood loss (ColoScreen) and general appearance.
  • this example provides an in vivo confirmation that the functional composition is effective in obtaining a stronger prevention of and faster and more potent recovery from intestinal inflammation and Disease Activity upon TNBS-induced colitis induction. Moreover, this finding clearly demonstrates that the preadaptation through the Collaborome strategy results in a more efficient activity as compared to the Assembly strategy
  • the chronic DSS model is a commonly used model for colitis that mimics some of the features of Crohn's disease, including weight loss and bloody diarrhea.
  • chronic DSS administration causes inflammation of the gut with typical architectural changes such as crypt distortion, (sub)mucosal infiltration of inflammatory cells and fibrosis, features found in patients with CD.
  • the composition containing Lactobacillus plantarum, Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila and Anaerostipes caccae, and produced through the "Collaborome” strategy (see example 1.3), is used to evaluate the beneficial effects upon evaluation in the chronic DSS model. Colitis is evoked in the animals by repeated administration of DSS in the drinking water (0.25% challenge). The experiment is performed over a total of 8 weeks, with 3 cycles of DSS administration and recovery.
  • DAI Disease Activity Index
  • Example 3.5 Mucositis model
  • Mucositis is a clinical term used to describe damage to mucous membranes after anticancer therapies. It occurs throughout the entire gastrointestinal tract (GT) (including the mouth) and genito-urinary tract, and to a lesser extent in other mucosal surfaces. Its severity and duration varies with the dose and the type of drug used.
  • GT gastrointestinal tract
  • the importance of mucositis is that it limits the dose of chemotherapy.
  • the GI crypt epithelium is particularly vulnerable to chemotherapeutic toxicity, with symptoms including nausea and vomiting, abdominal pain, distension, and diarrhea due to direct effects of the cytotoxics on the mucosa.
  • the 5-fluorouracyl (5FU)-induced gut mucositis rat model was established by Keefe et al. for assessment of the effects of chemotherapy on the GI tract and it is now one of the most extensively used models to investigate chemotherapy-induced mucositis in rats (Keefe 2004).
  • composition comprising Lactobacillus plantarum, Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Akkermansia muciniphila and Anaerostipes caccae, was used as basis for the experiment and produced via the "Collaborome" strategy of Example 1.3. Mucositis is induced by means of a single intraperitoneal dose of 5FU.
  • a total of 30 rats were randomly assigned to either a control or experimental group according to a specific time point. All rats in the experimental groups received a single intraperitoneal dose of 5FU (150mg 5FU/kg BW). Rats in the control groups received treatment with the solvent vehicle (dimethylsulphoxide). Subsequent to administration of the chemotherapy drugs, study endpoints such as mortality, diarrhea, and general clinical condition were assessed four times per 24 h period. Subgroups of the rats were killed by exsanguination and cervical dislocation at 24, 48, and 72 h following administration of the drug. Primary endpoints of interest were evolution of weight, diarrhea and general wellbeing (sickness score). Secondary endpoints included histology of intestinal samples and stool and gut mucosal microbiota analysis.
  • part of the rats were administered for 8 consecutive days with the composition by means of oral gavage. Preventive dosing started 5 days before the administration of 5FU and lasted for 3 days after 5FU administration or until rats were sacrificed. Control animals did not receive the composition.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nutrition Science (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Sustainable Development (AREA)
  • Physiology (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Polymers & Plastics (AREA)
  • Food Science & Technology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Dispersion Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)

Claims (13)

  1. Composition constituée essentiellement par des bactéries appartenant aux espèces Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum et Anaerostipes caccae ou appartenant aux espèces Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Akkermansia muciniphila, Lactobacillus plantarum et Anaerostipes caccae ou appartenant aux espèces Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum, Anaerostipes caccae, Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron et Bacteroides vulgatum.
  2. Composition selon la revendication 1, qui est destinée à être utilisée pour prévenir ou traiter des symptômes associés à un trouble gastro-intestinal.
  3. Composition destinée à être utilisée selon la revendication 2, dans laquelle ledit trouble gastro-intestinal est une interruption de la fonction de barrière des intestins, une diarrhée, une constipation, le syndrome du côlon irritable, une maladie inflammatoire de l'intestin, la maladie de Crohn, une colite ulcéreuse, la maladie cœliaque, une pochite, une mucosité, une infection intestinale, une dysbiose du microbiote intestinal et une quelconque combinaison de ceux-ci.
  4. Composition destinée à être utilisée selon les revendications 2 à 3, dans laquelle ledit trouble gastro-intestinal est prévenu ou traité via : a) une stimulation de la croissance et/ou de l'activité d'une bactérie bénéfique ou d'un nombre limité d'entre elles dans le tractus intestinal, b) une inhibition de la croissance et/ou de l'activité d'une bactérie pathogène ou d'un nombre limité d'entre elles dans le tractus intestinal, c) une augmentation relative de l'adhérence de bactéries non pathogènes à la muqueuse de la surface gastro-intestinale, d) une réduction de l'absorption incontrôlée d'antigènes, de bactéries pro-inflammatoires, ou de produits bactériens par les intestins, e) un apport d'une activité anti-inflammatoire au niveau de la surface intestinale, f) une augmentation du fonctionnement de la barrière intestinale, g) une production de métabolites bactériens ou h) une quelconque combinaison des paragraphes a) à g).
  5. Composition constituée essentiellement par des bactéries appartenant aux espèces Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum et Anaerostipes caccae ou appartenant aux espèces Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Akkermansia muciniphila, Lactobacillus plantarum et Anaerostipes caccae ou appartenant aux espèces Faecalibacieriurn prausnitzii, Butyricicoccus pullicaecorum, Roseburia inulinivorans, Roseburia hominis, Akkermansia muciniphila, Lactobacillus plantarum, Anaerostipes caccae, Escherichia coli, Enterococcus faecium, Lactobacillus mucosae, Bifidobacterium adolescentis, Bifidobacterium longum, Bacteroides thetaiotaomicron, Bacteroides vulgatum et un ou plusieurs probiotique(s).
  6. Composition selon les revendications 1 à 2 et 5, dans laquelle lesdites bactéries sont cultivées ensemble dans un fermenteur avant d'administrer ladite composition pour prévenir ou traiter lesdits troubles gastro-intestinaux.
  7. Composition selon la revendication 6, dans laquelle ledit fermenteur est un simulateur dynamique du tractus gastro-intestinal.
  8. Composition selon la revendication 1, dans laquelle lesdites bactéries sont choisies à partir de la liste des souches suivantes : Faecalibacterium prausnitzii LMG P-29362, Faecalibacterium prausnitzii DSMZ 17677, Butyricicoccus pullicaecorum LMG P-29360, Butyricicoccus pullicaecorum LMG24109, Roseburia inulinivorans LMG P-29365, Roseburia inulinivorans DSMZ 16841, Roseburia hominis LMG P-29364, Roseburia hominis DSMZ 16839, Akkermansia muciniphila LMG P-29361, Akkermansia muciniphila DSMZ 22959, Lactobacillus plantarum LMG P-29366, Lactobacillus plantarum ZJ316, Anaerostipes caccae LMG P-29359, Anaerostipes caccae DSMZ 14662 ou des souches présentant une identité de séquence d'au moins 97% avec les séquences d'ARNr16S d'au moins l'une desdites souches.
  9. Composition destinée à être utilisée selon les revendications 1 à 2 et 5 à 8, ladite composition étant une composition pharmaceutique formulée soit sous une forme administrée par voie rectale, soit sous une forme ingérable par voie orale.
  10. Composition selon la revendication 9, dans laquelle ladite forme ingérable par voie orale est une capsule, une microcapsule, un comprimé, une granule, une poudre, une pastille, une pilule, une suspension ou un sirop.
  11. Composition, selon la revendication 10, qui est incorporée dans un aliment, une boisson, un complément alimentaire ou une substance nutraceutique.
  12. Composition selon les revendications 1 à 2 et 5 à 11, ladite composition comprenant entre 105 et 1011 unités formant des colonies de bactéries.
  13. Réacteur comprenant une composition telle que définie dans l'une quelconque des revendications 1 à 2 et 5 à 12.
EP17703726.4A 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale Active EP3411052B8 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP23182773.4A EP4257194A3 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP19212983.1A EP3639834B1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
PL17703726T PL3411052T3 (pl) 2016-02-04 2017-02-03 Zastosowanie skupisk drobnoustrojów dla zdrowia ludzi i zwierząt
SI201730441T SI3411052T1 (sl) 2016-02-04 2017-02-03 Uporaba mikrobnih skupnosti za zdravje ljudi in živali
DK19212983.1T DK3639834T3 (da) 2016-02-04 2017-02-03 Anvendelse af mikrobielle samfund til human- og dyresundhed
RS20201197A RS60918B1 (sr) 2016-02-04 2017-02-03 Upotreba mikrobnih zajednica za zdravlje ljudi i životinja
HRP20201561TT HRP20201561T1 (hr) 2016-02-04 2020-09-30 Uporaba mikrobnih zajednica za zdravlje ljudi i životinja

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16154288 2016-02-04
PCT/EP2017/052422 WO2017134240A1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale

Related Child Applications (3)

Application Number Title Priority Date Filing Date
EP19212983.1A Division EP3639834B1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP19212983.1A Division-Into EP3639834B1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP23182773.4A Division EP4257194A3 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale

Publications (3)

Publication Number Publication Date
EP3411052A1 EP3411052A1 (fr) 2018-12-12
EP3411052B1 true EP3411052B1 (fr) 2020-07-08
EP3411052B8 EP3411052B8 (fr) 2020-09-30

Family

ID=55310719

Family Applications (3)

Application Number Title Priority Date Filing Date
EP17703726.4A Active EP3411052B8 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP23182773.4A Pending EP4257194A3 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP19212983.1A Active EP3639834B1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale

Family Applications After (2)

Application Number Title Priority Date Filing Date
EP23182773.4A Pending EP4257194A3 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale
EP19212983.1A Active EP3639834B1 (fr) 2016-02-04 2017-02-03 Utilisation de communautés microbiennes pour la santé humaine et animale

Country Status (24)

Country Link
US (7) US11096971B2 (fr)
EP (3) EP3411052B8 (fr)
JP (3) JP7016112B2 (fr)
KR (1) KR20180127972A (fr)
CN (2) CN108712906B (fr)
AU (2) AU2017216285B2 (fr)
BR (1) BR112018015097A2 (fr)
CA (1) CA3011083A1 (fr)
CY (1) CY1123954T1 (fr)
DK (2) DK3639834T3 (fr)
ES (2) ES2824536T3 (fr)
FI (1) FI3639834T3 (fr)
HK (1) HK1257070A1 (fr)
HR (1) HRP20231182T1 (fr)
HU (2) HUE051114T2 (fr)
IL (3) IL293486B2 (fr)
LT (2) LT3411052T (fr)
MX (1) MX2018009539A (fr)
PL (2) PL3639834T3 (fr)
PT (1) PT3639834T (fr)
RS (2) RS60918B1 (fr)
RU (2) RU2758387C2 (fr)
SI (2) SI3639834T1 (fr)
WO (1) WO2017134240A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3911161A4 (fr) * 2019-01-16 2022-09-14 Board of Regents, The University of Texas System Méthodes et compositions pour traiter une colite associée à un inhibiteur de point de contrôle immunitaire

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2964480A1 (fr) 2014-10-31 2016-05-06 Whole Biome Inc. Procedes et compositions se rapportant a un traitement microbien et au diagnostic de troubles
LT3411052T (lt) 2016-02-04 2021-01-11 Universiteit Gent Mikrobinių bendrijų naudojimas žmogaus ir gyvūno sveikatos pagerinimui
EP3423093A4 (fr) * 2016-03-04 2019-11-13 The Regents of The University of California Consortium microbien et utilisations de ce dernier
WO2018145082A1 (fr) * 2017-02-06 2018-08-09 New York University Méthodes et compositions pour traiter et diagnostiquer les cancers du pancréas
CN107384828B (zh) * 2017-08-22 2020-06-16 中国农业科学院农产品加工研究所 阿克曼氏粘细菌培养基及其制备方法
EP3675882A4 (fr) 2017-08-30 2021-07-28 Pendulum Therapeutics, Inc. Methodes et compositions pour le traitement de troubles associés au microbiome
WO2019098810A2 (fr) * 2017-11-20 2019-05-23 경희대학교 산학협력단 Nouvelles bactéries lactiques et leur utilisation
KR102250597B1 (ko) 2017-11-20 2021-05-11 경희대학교 산학협력단 신규 유산균 및 이의 용도
WO2019199895A1 (fr) 2018-04-10 2019-10-17 Siolta Therapeutics, Inc. Consortia microbiens
CN112292141A (zh) * 2018-04-13 2021-01-29 医学生命探索有限公司 产生长链双羧基脂肪酸(lcdfa)的微生物及其用途
WO2019211469A1 (fr) * 2018-05-04 2019-11-07 4D Pharma Research Limited Environnement intestinal simulé
CA3106315A1 (fr) * 2018-07-19 2020-01-23 Pendulum Therapeutics, Inc. Procedes et compositions pour prise de greffe microbienne
US11859214B1 (en) 2018-08-17 2024-01-02 The Government Of The United States, As Represented By The Secretary Of The Army Automated system for simulating the human lower gastrointestinal tract
EP3870691A4 (fr) * 2018-09-13 2022-09-07 Xbiome Inc. Procédés et compositions pour traiter des troubles gastro-intestinaux et inflammatoires
WO2020079026A1 (fr) * 2018-10-15 2020-04-23 Pharmabiome Ag Procédé de fabrication d'un consortium de souches bactériennes
CN110607262B (zh) * 2019-09-25 2022-03-25 君维安(武汉)生命科技有限公司 一种干预炎性肠炎的益生菌组合物及其筛选方法和应用
CN110448577B (zh) * 2019-09-26 2023-06-23 青岛农业大学 一种溃疡性结肠炎修复益生菌微胶囊制剂
CA3153884A1 (fr) 2019-10-07 2021-04-15 Nikole KIMES Compositions pharmaceutiques therapeutiques
WO2021072322A1 (fr) * 2019-10-09 2021-04-15 Mayo Foundation For Medical Education And Research Traitement de troubles gastro-intestinaux
GB201915144D0 (en) * 2019-10-18 2019-12-04 Multigerm Uk Entpr Ltd Method of promoting SCFA production by gut microbiota
CN110916192B (zh) * 2019-11-25 2021-04-13 垒途智能教科技术研究院江苏有限公司 一种具有防治儿童青少年自闭症的益生菌矿物粉及其应用
JP2023507204A (ja) 2019-12-23 2023-02-21 エボニック オペレーションズ ゲーエムベーハー グルテン分解のためのプロバイオティクス菌株を含む調製物
CN114657083A (zh) * 2019-12-24 2022-06-24 顾青 一种乳酸菌发酵乳
CN111117925B (zh) * 2020-01-13 2021-05-25 浙江大学 Anaerostipes sp B2131菌及其在炎症性肠病中的应用
CN111413503A (zh) * 2020-03-28 2020-07-14 浙江大学 骨桥蛋白作为靶分子在调节高脂饮食引起的肥胖中的应用
AU2021254477A1 (en) * 2020-04-08 2022-11-10 Megmilk Snow Brand Co., Ltd. Composition for improving gut microbiota
EP4132550A4 (fr) * 2020-04-08 2024-02-28 Psomagen, Inc. Composition et méthode pour un déséquilibre induisant un antibiotique dans le microbiote
KR102169794B1 (ko) * 2020-06-24 2020-10-27 주식회사 엔테로바이옴 신규한 피칼리박테리움 프로스니치 균주 eb-fpdk11 및 그의 용도
CN111938158B (zh) * 2020-08-18 2023-07-07 广东弘元普康医疗科技有限公司 一种防止肠道Akkermansia muciniphila菌丰度降低的组合物
GB202015687D0 (en) * 2020-10-02 2020-11-18 Microbiotica Ltd Therapeutic composition
CN113215020B (zh) * 2021-02-22 2022-06-17 中国科学院微生物研究所 罗氏菌mgb-2及其应用
CN113214976B (zh) * 2021-03-29 2023-04-14 中南大学 一种高效筛选嗜酸微生物的装置及其筛选方法
BE1029496B1 (fr) * 2021-08-19 2023-01-16 The Akkermansia Company Composition comprenant de l'Akkermansia muciniphila pasteurisée pour le traitement ou la prévention des troubles de la contractilité intestinale, en particulier des troubles de l'amplitude de la contractilité duodénale
AU2022426633A1 (en) 2021-12-27 2024-07-11 Mrm Health N.V. Microbial communities
CN114292781B (zh) * 2021-12-28 2023-08-01 中山大学 一种长双歧杆菌sysu-02及其应用
CN114304380A (zh) * 2022-01-13 2022-04-12 合肥河川生物医药科技有限公司 普拉梭菌在猪鸡上的应用
NL2032642B1 (en) * 2022-07-29 2024-02-06 Academisch Ziekenhuis Leiden Improvement of muscle mass and strength
WO2024049207A1 (fr) * 2022-08-31 2024-03-07 주식회사 바이오뱅크힐링 Nouvelle souche et vésicules qui en sont issues, et leurs utilisations anti-inflammatoires et antibactériennes
CN115737687A (zh) * 2022-10-16 2023-03-07 新疆医科大学第一附属医院 普拉梭菌在制备预防或/和治疗冠心病药物或/和保健品中的应用
WO2024123891A1 (fr) * 2022-12-07 2024-06-13 The University Of North Carolina At Chapel Hill Office Of Technology Commercialization Compositions de micro-organismes modifiés et leurs applications
CN116836880B (zh) * 2023-07-24 2023-11-28 中山大学 一种普氏梭杆菌及其应用
CN116925975B (zh) * 2023-08-18 2024-01-23 善恩康生物科技(苏州)有限公司 一种Akkermansia muciniphila及其产品和应用

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0307026D0 (en) * 2003-03-27 2003-04-30 Rowett Res Inst Bacterial supplement
EP2277524B1 (fr) * 2005-09-28 2015-11-11 Nordisk Rebalance A/S Traitement du syndrome inflammatoire de l'intestin par l'utilisation en tant qu'effecteurs thérapeutiques, de bactéries probiotiques et de céréales fermentées
CN101353633B (zh) * 2008-06-13 2011-12-28 浙江工商大学 Lactobacillus plantarum ZJ316、产生的抗菌肽及其制备与应用
GB0903016D0 (en) * 2009-02-23 2009-04-08 Univ Gent Method for alleviating intestinal problems and novel bacterial strains therefor
JP6006117B2 (ja) * 2009-11-12 2016-10-12 ネステク ソシエテ アノニム 腸内微生物叢バランス及び健康を促進するための栄養組成物
US8445226B2 (en) * 2010-02-01 2013-05-21 Microbios, Inc. Process and composition for the manufacture of a microbial-based product
US20120034198A1 (en) * 2010-08-04 2012-02-09 Microbios, Inc. Carriers for storage and delivery of biologics
WO2012142605A1 (fr) * 2011-04-15 2012-10-18 Samaritan Health Services Agent de déploiement de recolonisation rapide
CA2848762C (fr) 2011-09-14 2021-07-27 Queen's University At Kingston Procede de traitement de troubles de l'appareil gastro-intestinal
GB201117313D0 (en) * 2011-10-07 2011-11-16 Gt Biolog Ltd Bacterium for use in medicine
CN103082292B (zh) * 2011-11-02 2015-03-04 深圳华大基因研究院 罗斯氏菌(Roseburia)在治疗和预防肥胖相关疾病中的应用
WO2014075745A1 (fr) 2012-11-19 2014-05-22 Université Catholique de Louvain Utilisation d'akkermansia pour le traitement de troubles métaboliques
US8906668B2 (en) * 2012-11-23 2014-12-09 Seres Health, Inc. Synergistic bacterial compositions and methods of production and use thereof
SG10201704035TA (en) 2012-11-23 2017-06-29 Seres Therapeutics Inc Synergistic bacterial compositions and methods of production and use thereof
KR102222273B1 (ko) * 2013-02-04 2021-03-08 세레스 테라퓨틱스, 인코포레이티드 조성물 및 방법
BR112015020819A2 (pt) * 2013-03-05 2017-07-18 Academisch Ziekenhuis Groningen uso de faecalibacterium prausnitzii htf-f (dsm 26943) para supressão de inflamação
AU2014232370B2 (en) 2013-03-15 2018-11-01 Seres Therapeutics, Inc. Network-based microbial compositions and methods
US10058576B2 (en) * 2013-10-03 2018-08-28 The Trustees Of The University Of Pennsylvania Compositions and methods comprising a defined microbiome and methods of use thereof
KR102379658B1 (ko) * 2013-11-25 2022-03-28 세레스 테라퓨틱스, 인코포레이티드 상승적 박테리아 조성물 및 그것의 생산 방법 및 용도
EP2944696A1 (fr) 2014-05-13 2015-11-18 Evonik Degussa GmbH Procédé de production de composés organiques
CA2964480A1 (fr) * 2014-10-31 2016-05-06 Whole Biome Inc. Procedes et compositions se rapportant a un traitement microbien et au diagnostic de troubles
LT3411052T (lt) * 2016-02-04 2021-01-11 Universiteit Gent Mikrobinių bendrijų naudojimas žmogaus ir gyvūno sveikatos pagerinimui

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3911161A4 (fr) * 2019-01-16 2022-09-14 Board of Regents, The University of Texas System Méthodes et compositions pour traiter une colite associée à un inhibiteur de point de contrôle immunitaire

Also Published As

Publication number Publication date
IL293486A (en) 2022-08-01
CN116570631A (zh) 2023-08-11
US11096971B2 (en) 2021-08-24
RU2018131244A3 (fr) 2020-06-18
US11596658B2 (en) 2023-03-07
CY1123954T1 (el) 2022-03-24
CA3011083A1 (fr) 2017-08-10
DK3411052T3 (da) 2020-10-12
JP7016112B2 (ja) 2022-02-04
RS60918B1 (sr) 2020-11-30
EP3639834A1 (fr) 2020-04-22
BR112018015097A2 (pt) 2018-12-26
IL260337A (en) 2018-08-30
WO2017134240A1 (fr) 2017-08-10
LT3639834T (lt) 2023-11-10
PT3639834T (pt) 2023-10-12
CN108712906B (zh) 2023-06-30
DK3639834T3 (da) 2023-10-09
US20210353694A1 (en) 2021-11-18
ES2960781T3 (es) 2024-03-06
EP3411052B8 (fr) 2020-09-30
KR20180127972A (ko) 2018-11-30
US20230028557A1 (en) 2023-01-26
JP2024028833A (ja) 2024-03-05
US20220072071A1 (en) 2022-03-10
IL283594A (en) 2021-07-29
JP7460979B2 (ja) 2024-04-03
JP2019511563A (ja) 2019-04-25
IL260337B (en) 2021-06-30
AU2017216285B2 (en) 2023-10-05
US20190015465A1 (en) 2019-01-17
LT3411052T (lt) 2021-01-11
RU2758387C2 (ru) 2021-10-28
US20220023357A1 (en) 2022-01-27
US20230045069A1 (en) 2023-02-09
US20220040245A1 (en) 2022-02-10
PL3639834T3 (pl) 2024-01-29
RU2021130638A (ru) 2021-11-18
IL293486B2 (en) 2024-04-01
HUE051114T2 (hu) 2021-01-28
EP4257194A3 (fr) 2023-12-20
JP2022058615A (ja) 2022-04-12
RU2018131244A (ru) 2020-03-04
AU2023248066A1 (en) 2023-11-02
FI3639834T3 (fi) 2023-09-27
IL293486B1 (en) 2023-12-01
IL283594B (en) 2022-06-01
ES2824536T3 (es) 2021-05-12
EP3411052A1 (fr) 2018-12-12
HK1257070A1 (zh) 2019-10-11
MX2018009539A (es) 2019-02-11
PL3411052T3 (pl) 2021-03-08
EP3639834B1 (fr) 2023-07-12
SI3639834T1 (sl) 2024-02-29
SI3411052T1 (sl) 2021-01-29
US11491196B2 (en) 2022-11-08
HRP20231182T1 (hr) 2024-01-05
HUE063584T2 (hu) 2024-01-28
RS64782B1 (sr) 2023-11-30
CN108712906A (zh) 2018-10-26
EP4257194A2 (fr) 2023-10-11
US11633440B2 (en) 2023-04-25
AU2017216285A1 (en) 2018-07-26

Similar Documents

Publication Publication Date Title
US11491196B2 (en) Use of microbial communities for human and animal health
US11701396B2 (en) Treatment of Clostridium difficile infection
Saarela et al. Gut bacteria and health foods—the European perspective
TWI744479B (zh) 包含雙歧桿菌的組合物及其製備方法
GUIDESI INTEGRATED APPROACH TO THE SELECTION OF NEW PROBIOTICS FOR HUMAN APPLICATION

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180626

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20191014

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
RIN1 Information on inventor provided before grant (corrected)

Inventor name: VAN DE WIELE, TOM

Inventor name: POSSEMIERS, SAM

Inventor name: VAN DEN ABEELE, PIETER

Inventor name: GOTTARDI, DAVIDE

Inventor name: BOLCA, SELIN

Inventor name: SCHEIRLINCK, ILSE

Inventor name: MARZORATI, MASSIMO

INTG Intention to grant announced

Effective date: 20200212

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1287716

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200715

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602017019338

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602017019338

Country of ref document: DE

Owner name: UNIVERSITEIT GENT, BE

Free format text: FORMER OWNERS: PRODIGEST BVBA, ZWIJNAARDE, BE; UNIVERSITEIT GENT, GENT, BE

Ref country code: DE

Ref legal event code: R081

Ref document number: 602017019338

Country of ref document: DE

Owner name: MICROBIAL RESOURCE MANAGEMENT HEALTH NV, BE

Free format text: FORMER OWNERS: PRODIGEST BVBA, ZWIJNAARDE, BE; UNIVERSITEIT GENT, GENT, BE

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: UNIVERSITEIT GENT

Owner name: MICROBIAL RESOURCE MANAGEMENT HEALTH NV

REG Reference to a national code

Ref country code: CH

Ref legal event code: PK

Free format text: BERICHTIGUNG B8

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20201561T

Country of ref document: HR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3411052

Country of ref document: PT

Date of ref document: 20201008

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20200929

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20201006

Ref country code: FI

Ref legal event code: FGE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20200708

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20200402787

Country of ref document: GR

Effective date: 20201116

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E019886

Country of ref document: EE

Effective date: 20201006

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 35714

Country of ref document: SK

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20201561

Country of ref document: HR

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E051114

Country of ref document: HU

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20201561

Country of ref document: HR

Payment date: 20210121

Year of fee payment: 5

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602017019338

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: PD

Owner name: MICROBIAL RESOURCE MANAGEMENT HEALTH NV; BE

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: UNIVERSITEIT GENT

Effective date: 20210331

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2824536

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20210512

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20210409

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200708

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210203

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20201561

Country of ref document: HR

Payment date: 20220127

Year of fee payment: 6

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20201561

Country of ref document: HR

Payment date: 20230130

Year of fee payment: 7

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1287716

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200708

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230630

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AL

Payment date: 20230211

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20231221

Year of fee payment: 8

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20201561

Country of ref document: HR

Payment date: 20240125

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20230123

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20240221

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IS

Payment date: 20240212

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LT

Payment date: 20240123

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20240220

Year of fee payment: 8

Ref country code: NL

Payment date: 20240219

Year of fee payment: 8

Ref country code: ES

Payment date: 20240326

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20240220

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SM

Payment date: 20240129

Year of fee payment: 8

Ref country code: RO

Payment date: 20240125

Year of fee payment: 8

Ref country code: HU

Payment date: 20240222

Year of fee payment: 8

Ref country code: FI

Payment date: 20240219

Year of fee payment: 8

Ref country code: EE

Payment date: 20240215

Year of fee payment: 8

Ref country code: DE

Payment date: 20240219

Year of fee payment: 8

Ref country code: CZ

Payment date: 20240126

Year of fee payment: 8

Ref country code: CY

Payment date: 20240202

Year of fee payment: 8

Ref country code: BG

Payment date: 20240222

Year of fee payment: 8

Ref country code: GB

Payment date: 20240219

Year of fee payment: 8

Ref country code: CH

Payment date: 20240301

Year of fee payment: 8

Ref country code: SK

Payment date: 20240129

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SI

Payment date: 20240125

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20240202

Year of fee payment: 8

Ref country code: SE

Payment date: 20240219

Year of fee payment: 8

Ref country code: RS

Payment date: 20240126

Year of fee payment: 8

Ref country code: PL

Payment date: 20240125

Year of fee payment: 8

Ref country code: NO

Payment date: 20240222

Year of fee payment: 8

Ref country code: MT

Payment date: 20240124

Year of fee payment: 8

Ref country code: LV

Payment date: 20240214

Year of fee payment: 8

Ref country code: IT

Payment date: 20240228

Year of fee payment: 8

Ref country code: HR

Payment date: 20240125

Year of fee payment: 8

Ref country code: FR

Payment date: 20240221

Year of fee payment: 8

Ref country code: DK

Payment date: 20240223

Year of fee payment: 8

Ref country code: BE

Payment date: 20240219

Year of fee payment: 8